1
|
Li R, Song M, Zheng Y, Zhang J, Zhang S, Fan X. Naoxueshu oral liquid promotes hematoma absorption by targeting CD36 in M2 microglia via TLR4/MyD88/NF-κB signaling pathway in rats with intracerebral hemorrhage. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117116. [PMID: 37659762 DOI: 10.1016/j.jep.2023.117116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Intracerebral hemorrhage (ICH) is a major public health issue that leads to elevated rates of death and disability and has few proven treatments. Naoxueshu oral liquid (NXS), a TCM patent drug, is widely used in patients with ICH. Although a series of clinical studies have confirmed the efficacy and safety of NXS, the underlying mechanism of hematoma absorption is unclear. AIM OF THE STUDY Our work aimed to elucidate the effect and mechanism of NXS on hematoma absorption in rats with ICH. MATERIALS AND METHODS Induction of ICH model in the rats with intracerebral injection of collagenase VII, followed by treatment with NXS and Edaravone as a control neuroprotection medication. Neural functional recovery was assessed using mNSS, foot fault test, corner test, forelimb grip-traction test, and adhesive removal test. Hematoma absorption was assessed by the spectrophotometric hemoglobin assay with Drabkin's reagent. The protein expression of CD36, M2 microglia marker (CD206 and YM-1) and TLR4/MyD88/NF-κB pathway related proteins were determined by Western blot and immunofluorescence. RESULTS NXS could significantly ameliorate the ICH recovery of neural and locomotor function as well as reduce hemorrhage volume. NXS could increase the expression of CD36 expressed in M2 microglia and promote M2 microglia polarization. Simultaneously, NXS significantly suppressed protein expressions of TLR4, MyD88, and NF-κB following ICH in rats. The results indicated that lipopolysaccharide (LPS), TLR4 specific agonist, could partially reverse the change in ICH rats administrated with NXS. CONCLUSIONS NXS promotes hematoma absorption by targeting CD36 expression in M2 microglia via TLR4/MyD88/NF-κB signaling pathway in rats with ICH. Collectively, current research provides a novel theoretical basis for the clinical application of NXS.
Collapse
Affiliation(s)
- Ruoqi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Meiying Song
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yingyi Zheng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jiaxue Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
2
|
Kawamura T, Singh Mallah G, Ardalan M, Chumak T, Svedin P, Jonsson L, Jabbari Shiadeh SM, Goretta F, Ikeda T, Hagberg H, Sandberg M, Mallard C. Therapeutic Effect of Nicotinamide Mononucleotide for Hypoxic-Ischemic Brain Injury in Neonatal Mice. ASN Neuro 2023; 15:17590914231198983. [PMID: 37787108 PMCID: PMC10548811 DOI: 10.1177/17590914231198983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 10/04/2023] Open
Abstract
SUMMARY STATEMENT Neonatal hypoxia-ischemia reduces nicotinamide adenine dinucleotide (NAD+) and SIRT6 levels in the injured hippocampus.Hippocampal high mobility group box-1 (HMGB1) release is significantly increased after neonatal hypoxia-ischemia.Nicotinamide mononucleotide (NMN) treatment normalizes hippocampal NAD+ and SIRT6 levels, with significant decrease in caspase-3 activity and HMGB1 release.NMN improves early developmental behavior, as well as motor and memory function.
Collapse
Affiliation(s)
- Takuya Kawamura
- Institute of Neuroscience and Physiology, Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Mie University, Tsu, Japan
| | - Gagandeep Singh Mallah
- Institute of Neuroscience and Physiology, Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maryam Ardalan
- Institute of Neuroscience and Physiology, Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tetyana Chumak
- Institute of Neuroscience and Physiology, Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Svedin
- Institute of Neuroscience and Physiology, Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lina Jonsson
- Institute of Neuroscience and Physiology, Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Seyedeh Marziyeh Jabbari Shiadeh
- Institute of Neuroscience and Physiology, Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fanny Goretta
- Institute of Neuroscience and Physiology, Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Mie University, Tsu, Japan
| | - Henrik Hagberg
- Centre of Perinatal Medicine and Health, Institute of Clinical Sciences, Gothenburg, Sweden
| | - Mats Sandberg
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
3
|
N-Acetylcysteine Administration Attenuates Sensorimotor Impairments Following Neonatal Hypoxic-Ischemic Brain Injury in Rats. Int J Mol Sci 2022; 23:ijms232416175. [PMID: 36555816 PMCID: PMC9783020 DOI: 10.3390/ijms232416175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hypoxic ischemic (HI) brain injury that occurs during neonatal period has been correlated with severe neuronal damage, behavioral deficits and infant mortality. Previous evidence indicates that N-acetylcysteine (NAC), a compound with antioxidant action, exerts a potential neuroprotective effect in various neurological disorders including injury induced by brain ischemia. The aim of the present study was to investigate the role of NAC as a potential therapeutic agent in a rat model of neonatal HI brain injury and explore its long-term behavioral effects. To this end, NAC (50 mg/kg/dose, i.p.) was administered prior to and instantly after HI, in order to evaluate hippocampal and cerebral cortex damage as well as long-term functional outcome. Immunohistochemistry was used to detect inducible nitric oxide synthase (iNOS) expression. The results revealed that NAC significantly alleviated sensorimotor deficits and this effect was maintained up to adulthood. These improvements in functional outcome were associated with a significant decrease in the severity of brain damage. Moreover, NAC decreased the short-term expression of iNOS, a finding implying that iNOS activity may be suppressed and that through this action NAC may exert its therapeutic action against neonatal HI brain injury.
Collapse
|
4
|
Shi Z, Luo K, Jani S, February M, Fernandes N, Venkatesh N, Sharif N, Tan S. Mimicking partial to total placental insufficiency in a rabbit model of cerebral palsy. J Neurosci Res 2022; 100:2138-2153. [PMID: 34173261 PMCID: PMC8709884 DOI: 10.1002/jnr.24901] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/24/2021] [Accepted: 05/24/2021] [Indexed: 01/07/2023]
Abstract
All placental abruptions begin as partial abruptions, which sometimes manifest as fetal bradycardia. The progression from partial to total abruption was mimicked by a new rabbit model of placental insufficiency, and we compared it, with sufficient statistical power, with the previous model mimicking total placental abruption. The previous model uses total uterine ischemia at E22 or E25 (70% or 79% term, respectively), in pregnant New Zealand white rabbits for 40 min (Full H-I). The new model, Partial+Full H-I, added a 30-min partial ischemia before the 40-min total ischemia. Fetuses were delivered either at E31.5 (full term) vaginally for neurobehavior testing, or by C-section at E25 for ex vivo brain cell viability evaluation. The onset of fetal bradycardia was within the first 2 min of either H-I protocol. There was no difference between Full H-I (n = 442 for E22, 312 for E25) and Partial+Full H-I (n = 154 and 80) groups in death or severely affected kits at E22 (76% vs. 79%) or at E25 (66% vs. 64%), or normal kits at E22 or E25, or any of the individual newborn neurobehavioral tests at any age. No sex differences were found. Partial+Full H-I (n = 6) showed less cell viability than Full H-I (n = 8) at 72-hr ex vivo in the brain regions studied. Partial+Full H-I insult produced similar cerebral palsy phenotype as our previous Full H-I model in a sufficiently powered study and may be more suitable for testing of potential neuroprotectants.
Collapse
Affiliation(s)
- Zhongjie Shi
- Department of Pediatrics, Children’s Hospital of Michigan, Detroit, MI
| | - Kehuan Luo
- Department of Pediatrics, Children’s Hospital of Michigan, Detroit, MI
| | - Sanket Jani
- Department of Pediatrics, Children’s Hospital of Michigan, Detroit, MI
| | - Melissa February
- Department of Pediatrics, Children’s Hospital of Michigan, Detroit, MI
| | - Nithi Fernandes
- Department of Pediatrics, Children’s Hospital of Michigan, Detroit, MI
| | | | | | - Sidhartha Tan
- Department of Pediatrics, Children’s Hospital of Michigan, Detroit, MI
| |
Collapse
|
5
|
Jones NM, Nathanson AD, Chell S, DeAngelis E, Whelan G, Willé D, Cheng K. The prolyl hydroxylase inhibitor GSK1120360A reduces early brain injury, but protection is not maintained in a neonatal rat model of hypoxic ischaemic encephalopathy. Int J Dev Neurosci 2022; 82:423-435. [PMID: 35662244 DOI: 10.1002/jdn.10199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/10/2022] [Accepted: 05/31/2022] [Indexed: 11/06/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) in newborns is associated with high morbidity and mortality, with many babies suffering long-term neurological deficits. Currently, treatment options are limited to therapeutic hypothermia, which is not appropriate for use in all babies. Previous studies have shown protective effects of increasing the transcription factor-hypoxia-inducible factor-1 (HIF-1) in animal models, by using mild hypoxia or compounds that act as prolyl hydroxylase inhibitors (PHIs). Here, we aimed to examine the neuroprotective actions of an orally active, small molecule PHI, GSK1120360A in a neonatal rat model of hypoxia-ischemia (HI) compared to another PHI, desferrioxamine (DFX). Sprague-Dawley rats underwent HI surgery on postnatal day 7 (P7), where unilateral carotid artery occlusion was performed followed by hypoxia (8% oxygen, 3 h). Initial testing showed that GSK1120360A and erythropoietin levels were detectable in plasma at 6 h following oral exposure to GSK1120360A. For the short-term neuroprotection study, pups were assigned to receive either saline (s.c), desferrioxamine (DFX-200 mg/kg, s.c), methylcellulose (1%, oral) or GSK1120360A (30 mg/kg, oral) immediately after HI. Histological analysis showed that GSK1120360A in this setting reduced brain injury size 7 days after HI, compared to the methylcellulose vehicle control group. DFX had no significant effect on injury size compared to saline group at the same 7 day timepoint. In the long-term neuroprotection study, pups were randomly assigned to be administered methylcellulose (1%, oral) or GSK1120360A (30 mg/kg, oral) immediately after HI. On P42, rats underwent behavioural testing using the forelimb grip strength, grid walking and novel object recognition tasks, and brains were collected for histological analysis. Long-term behavioural deficits were observed in grid walking, grip strength and novel object recognition tests after HI which were not improved in the GSK1120360A treatment group compared to the methylcellulose group. Similarly, there was no improvement in injury size on P42 in the GSK1120360A study group compared to the methylcellulose group. Here, we have shown that GSK1120360A can reduce brain injury at 7 days but that this neuroprotective benefit is not maintained when examined at 5 weeks after HI.
Collapse
Affiliation(s)
- Nicole M Jones
- Department of Pharmacology, School of Medical Sciences, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Anton D Nathanson
- Department of Pharmacology, School of Medical Sciences, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Simon Chell
- Medicines Research Centre, GlaxoSmithKline, Stevenage, UK
| | | | - Greg Whelan
- Medicines Research Centre, GlaxoSmithKline, Stevenage, UK
| | - David Willé
- Medicines Research Centre, GlaxoSmithKline, Stevenage, UK
| | | |
Collapse
|
6
|
Investigation on the potential targets of Astragaloside IV against intracerebral hemorrhage based on network pharmacology and experimental validation. Bioorg Chem 2022; 127:105975. [PMID: 35728292 DOI: 10.1016/j.bioorg.2022.105975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/31/2022] [Accepted: 06/14/2022] [Indexed: 11/23/2022]
Abstract
Intracerebral hemorrhage (ICH) is a life-threatening type of stroke that affects millions of individuals worldwide. Astragaloside IV (AS-IV), the main bioactive ingredient in Radix Astragali, has been linked to a variety of pharmacologic actions, including stroke. However, the effects and potential mechanisms of AS-IV on hematoma absorption after ICH are still unknown. The study aims to identify potential targets and regulation mechanisms of AS-IV on hematoma absorption after ICH. Network pharmacology, molecular docking, pharmacodynamic study, and western blot were used in this study to explore the potential mechanisms. The results showed that AS-IV could improve the hematoma absorption and neurological outcomes in collagenase VII induced rat ICH models. Molecular docking results had shown that PI3K and AKT were the potential targets of AS-IV against ICH. The experimental validation showed that AS-IV could reduce phosphorylation expression of PI3K and AKT, thereby inhibiting the NF-κB and increasing CD36 expression. This study demonstrated that AS-IV could play a critical role on hematoma absorption after ICH by regulating the PI3K/AKT signaling pathway and promoting CD36 phagocytosis, which provided a new thought for the drug development of ICH.
Collapse
|
7
|
Brégère C, Schwendele B, Radanovic B, Guzman R. Microglia and Stem-Cell Mediated Neuroprotection after Neonatal Hypoxia-Ischemia. Stem Cell Rev Rep 2022; 18:474-522. [PMID: 34382141 PMCID: PMC8930888 DOI: 10.1007/s12015-021-10213-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2021] [Indexed: 12/14/2022]
Abstract
Neonatal hypoxia-ischemia encephalopathy (HIE) refers to a brain injury in term infants that can lead to death or lifelong neurological deficits such as cerebral palsy (CP). The pathogenesis of this disease involves multiple cellular and molecular events, notably a neuroinflammatory response driven partly by microglia, the brain resident macrophages. Treatment options are currently very limited, but stem cell (SC) therapy holds promise, as beneficial outcomes are reported in animal studies and to a lesser degree in human trials. Among putative mechanisms of action, immunomodulation is considered a major contributor to SC associated benefits. The goal of this review is to examine whether microglia is a cellular target of SC-mediated immunomodulation and whether the recruitment of microglia is linked to brain repair. We will first provide an overview on microglial activation in the rodent model of neonatal HI, and highlight its sensitivity to developmental age. Two complementary questions are then addressed: (i) do immune-related treatments impact microglia and provide neuroprotection, (ii) does stem cell treatment modulates microglia? Finally, the immune-related findings in patients enrolled in SC based clinical trials are discussed. Our review points to an impact of SCs on the microglial phenotype, but heterogeneity in experimental designs and methodological limitations hamper our understanding of a potential contribution of microglia to SC associated benefits. Thorough analyses of the microglial phenotype are warranted to better address the relevance of the neuroimmune crosstalk in brain repair and improve or advance the development of SC protocols in humans.
Collapse
Affiliation(s)
- Catherine Brégère
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland
| | - Bernd Schwendele
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland
| | - Boris Radanovic
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
8
|
da Conceição Pereira S, Manhães-de-Castro R, Visco DB, de Albuquerque GL, da Silva Calado CMS, da Silva Souza V, Toscano AE. Locomotion is impacted differently according to the perinatal brain injury model: Meta-analysis of preclinical studies with implications for cerebral palsy. J Neurosci Methods 2021; 360:109250. [PMID: 34116077 DOI: 10.1016/j.jneumeth.2021.109250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/03/2021] [Accepted: 06/05/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Different approaches to reproduce cerebral palsy (CP) in animals, contribute to the knowledge of the pathophysiological mechanism of this disease and provide a basis for the development of intervention strategies. Locomotion and coordination are the main cause of disability in CP, however, few studies highlight the quantitative differences of CP models, on locomotion parameters, considering the methodologies to cause brain lesions in the perinatal period. METHODS Studies with cerebral palsy animal models that assess locomotion parameters were systematically retrieved from Medline/PubMed, SCOPUS, LILACS, and Web of Science. Methodological evaluation of included studies and quantitative assessment of locomotion parameters were performed after eligibility screening. RESULTS CP models were induced by hypoxia-ischemia (HI), Prenatal ischemia (PI), lipopolysaccharide inflammation (LPS), intraventricular haemorrhage (IVH), anoxia (A), sensorimotor restriction (SR), and a combination of different models. Overall, 63 studies included in qualitative synthesis showed a moderate quality of evidence. 16 studies were included in the quantitative meta-analysis. Significant reduction was observed in models that combined LPS with HI related to distance traveled (SMD -7.24 95 % CI [-8.98, -5.51], Z = 1.18, p < 0.00001) and LPS with HI or anoxia with sensory-motor restriction (SMD -6.01, 95 % CI [-7.67, -4.35], Z = 7.11), or IVH (SMD -4.91, 95 % CI [-5.84, -3.98], Z = 10.31, p < 0.00001) related to motor coordination. CONCLUSION The combination of different approaches to reproduce CP in animals causes greater deficits in locomotion and motor coordination from the early stages of life to adulthood. These findings contribute to methodological refinement, reduction, and replacement in animal experimentation, favoring translational purposes.
Collapse
Affiliation(s)
- Sabrina da Conceição Pereira
- Posgraduate Program in Neuropsychiatry and Behavior Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Raul Manhães-de-Castro
- Posgraduate Program in Neuropsychiatry and Behavior Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Postgraduate Program in Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Diego Bulcão Visco
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Postgraduate Program in Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | | | - Vanessa da Silva Souza
- Posgraduate Program in Neuropsychiatry and Behavior Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Ana Elisa Toscano
- Posgraduate Program in Neuropsychiatry and Behavior Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Postgraduate Program in Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Department of Nursing, CAV, Federal University of Pernambuco, Vitória de Santo Antão, Pernambuco, Brazil.
| |
Collapse
|
9
|
Kagan BJ, Ermine CM, Frausin S, Parish CL, Nithianantharajah J, Thompson LH. Focal Ischemic Injury to the Early Neonatal Rat Brain Models Cognitive and Motor Deficits with Associated Histopathological Outcomes Relevant to Human Neonatal Brain Injury. Int J Mol Sci 2021; 22:ijms22094740. [PMID: 33947043 PMCID: PMC8124303 DOI: 10.3390/ijms22094740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/08/2023] Open
Abstract
Neonatal arterial ischemic stroke is one of the more severe birth complications. The injury can result in extensive neurological damage and is robustly associated with later diagnoses of cerebral palsy (CP). An important part of efforts to develop new therapies include the on-going refinement and understanding of animal models that capture relevant clinical features of neonatal brain injury leading to CP. The potent vasoconstrictor peptide, Endothelin-1 (ET-1), has previously been utilised in animal models to reduce local blood flow to levels that mimic ischemic stroke. Our previous work in this area has shown that it is an effective and technically simple approach for modelling ischemic injury at very early neonatal ages, resulting in stable deficits in motor function. Here, we aimed to extend this model to also examine the impact on cognitive function. We show that focal delivery of ET-1 to the cortex of Sprague Dawley rats on postnatal day 0 (P0) resulted in impaired learning in a touchscreen-based test of visual discrimination and correlated with important clinical features of CP including damage to large white matter structures.
Collapse
|
10
|
Dos Santos TM, Ramires Júnior OV, Alves VS, Coutinho-Silva R, Savio LEB, Wyse ATS. Hyperhomocysteinemia alters cytokine gene expression, cytochrome c oxidase activity and oxidative stress in striatum and cerebellum of rodents. Life Sci 2021; 277:119386. [PMID: 33774024 DOI: 10.1016/j.lfs.2021.119386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/04/2021] [Accepted: 03/15/2021] [Indexed: 12/13/2022]
Abstract
AIMS Homocysteine has been linked to neurodegeneration and motor function impairments. In the present study, we evaluate the effect of chronic mild hyperhomocysteinemia on the motor behavior (motor coordination, functional performance, and muscular force) and biochemical parameters (oxidative stress, energy metabolism, gene expression and/or protein abundance of cytokine related to the inflammatory pathways and acetylcholinesterase) in the striatum and cerebellum of Wistar male rats. MAIN METHODS Rodents were submitted to one injection of homocysteine (0.03 μmol Hcy/g of body weight) between 30th and 60th postnatal days twice a day. After hyperhomocysteinemia induction, rats were submitted to horizontal ladder walking, beam balance, suspension, and vertical pole tests and/or euthanized to brain dissection for biochemical and molecular assays. KEY FINDINGS Chronic mild hyperhomocysteinemia did not alter motor function, but induced oxidative stress and impaired mitochondrial complex IV activity in both structures. In the striatum, hyperhomocysteinemia decreased TNF-α gene expression and increased IL-1β gene expression and acetylcholinesterase activity. In the cerebellum, hyperhomocysteinemia increased gene expression of TNF-α, IL-1β, IL-10, and TGF-β, while the acetylcholinesterase activity was decreased. In both structures, hyperhomocysteinemia decreased acetylcholinesterase protein abundance without altering total p-NF-κB, NF-κB, Nrf-2, and cleaved caspase-3. SIGNIFICANCE Chronic mild hyperhomocysteinemia compromises several biochemical/molecular parameters, signaling pathways, oxidative stress, and chronic inflammation in the striatum and cerebellum of rats without impairing motor function. These alterations may be related to the mechanisms in which hyperhomocysteinemia has been linked to movement disorders later in life and neurodegeneration.
Collapse
Affiliation(s)
- Tiago Marcon Dos Santos
- Wyse's Lab, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil
| | - Osmar Vieira Ramires Júnior
- Wyse's Lab, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil
| | - Vinícius Santos Alves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro - UFRJ, Av. Carlos Chagas Filho, 373, CCS, Ilha do Fundão, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Robson Coutinho-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro - UFRJ, Av. Carlos Chagas Filho, 373, CCS, Ilha do Fundão, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Luiz Eduardo Baggio Savio
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro - UFRJ, Av. Carlos Chagas Filho, 373, CCS, Ilha do Fundão, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Angela T S Wyse
- Wyse's Lab, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil.
| |
Collapse
|
11
|
Bradford A, Hernandez M, Kearney E, Theriault L, Lim YP, Stonestreet BS, Threlkeld SW. Effects of Juvenile or Adolescent Working Memory Experience and Inter-Alpha Inhibitor Protein Treatment after Neonatal Hypoxia-Ischemia. Brain Sci 2020; 10:E999. [PMID: 33348631 PMCID: PMC7765798 DOI: 10.3390/brainsci10120999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/04/2020] [Accepted: 12/15/2020] [Indexed: 12/24/2022] Open
Abstract
Hypoxic-Ischemic (HI) brain injury in the neonate contributes to life-long cognitive impairment. Early diagnosis and therapeutic interventions are critical but limited. We previously reported in a rat model of HI two interventional approaches that improve cognitive and sensory function: administration of Inter-alpha Inhibitor Proteins (IAIPs) and early experience in an eight-arm radial water maze (RWM) task. Here, we expanded these studies to examine the combined effects of IAIPs and multiple weeks of RWM assessment beginning with juvenile or adolescent rats to evaluate optimal age windows for behavioral interventions. Subjects were divided into treatment groups; HI with vehicle, sham surgery with vehicle, and HI with IAIPs, and received either juvenile (P31 initiation) or adolescent (P52 initiation) RWM testing, followed by adult retesting. Error rates on the RWM decreased across weeks for all conditions. Whereas, HI injury impaired global performance as compared to shams. IAIP-treated HI subjects tested as juveniles made fewer errors as compared to their untreated HI counterparts. The juvenile group made significantly fewer errors on moderate demand trials and showed improved retention as compared to the adolescent group during the first week of adult retesting. Together, results support and extend our previous findings that combining behavioral and anti-inflammatory interventions in the presence of HI improves subsequent learning performance. Results further indicate sensitive periods for behavioral interventions to improve cognitive outcomes. Specifically, early life cognitive experience can improve long-term learning performance even in the presence of HI injury. Results from this study provide insight into typical brain development and the impact of developmentally targeted therapeutics and task-specific experience on subsequent cognitive processing.
Collapse
Affiliation(s)
- Aaron Bradford
- Neuroscience Program, School of Health Sciences, Regis College, 235 Wellesley Street, Weston, MA 02493, USA; (A.B.); (M.H.); (E.K.); (L.T.)
| | - Miranda Hernandez
- Neuroscience Program, School of Health Sciences, Regis College, 235 Wellesley Street, Weston, MA 02493, USA; (A.B.); (M.H.); (E.K.); (L.T.)
| | - Elaine Kearney
- Neuroscience Program, School of Health Sciences, Regis College, 235 Wellesley Street, Weston, MA 02493, USA; (A.B.); (M.H.); (E.K.); (L.T.)
| | - Luke Theriault
- Neuroscience Program, School of Health Sciences, Regis College, 235 Wellesley Street, Weston, MA 02493, USA; (A.B.); (M.H.); (E.K.); (L.T.)
| | - Yow-Pin Lim
- ProThera Biologics, Inc., 349 Eddy Street, Providence, RI 02903, USA;
- Department of Pathology and Laboratory Medicine, The Alpert Medical School of Brown University, 222 Richmond Street, Providence, RI 02903, USA
| | - Barbara S. Stonestreet
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, 101 Dudley Street, Providence, RI 02905, USA;
| | - Steven W. Threlkeld
- Neuroscience Program, School of Health Sciences, Regis College, 235 Wellesley Street, Weston, MA 02493, USA; (A.B.); (M.H.); (E.K.); (L.T.)
| |
Collapse
|
12
|
TLR4 inhibition ameliorates mesencephalic substantia nigra injury in neonatal rats exposed to lipopolysaccharide via regulation of neuro-immunity. Brain Res Bull 2020; 165:90-96. [DOI: 10.1016/j.brainresbull.2020.09.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 02/02/2023]
|
13
|
Feng L, Han CX, Cao SY, Zhang HM, Wu GY. Deficits in motor and cognitive functions in an adult mouse model of hypoxia-ischemia induced stroke. Sci Rep 2020; 10:20646. [PMID: 33244072 PMCID: PMC7692481 DOI: 10.1038/s41598-020-77678-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 11/13/2020] [Indexed: 11/25/2022] Open
Abstract
Ischemic strokes cause devastating brain damage and functional deficits with few treatments available. Previous studies have shown that the ischemia-hypoxia rapidly induces clinically similar thrombosis and neuronal loss, but any resulting behavioral changes are largely unknown. The goal of this study was to evaluate motor and cognitive deficits in adult HI mice. Following a previously established procedure, HI mouse models were induced by first ligating the right common carotid artery and followed by hypoxia. Histological data showed significant long-term neuronal losses and reactive glial cells in the ipsilateral striatum and hippocampus of the HI mice. Whereas the open field test and the rotarod test could not reliably distinguish between the sham and HI mice, in the tapered beam and wire-hanging tests, the HI mice showed short-term and long-term deficits, as evidenced by the increased number of foot faults and decreased hanging time respectively. In cognitive tests, the HI mice swam longer distances and needed more time to find the platform in the Morris water maze test and showed shorter freezing time in fear contextual tests after fear training. In conclusion, this study demonstrates that adult HI mice have motor and cognitive deficits and could be useful models for preclinical stroke research.
Collapse
Affiliation(s)
- Li Feng
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China.
| | - Chun-Xia Han
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Shu-Yu Cao
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - He-Ming Zhang
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China.
| | - Gang-Yi Wu
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| |
Collapse
|
14
|
Adams KV, Mahmud N, Green-Holland M, Vonderwalde I, Umebayashi D, Sachewsky N, Coles BL, van der Kooy D, Morshead CM. Constraint-induced movement therapy promotes motor recovery after neonatal stroke in the absence of neural precursor activation. Eur J Neurosci 2020; 53:1334-1349. [PMID: 33010080 DOI: 10.1111/ejn.14993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 09/08/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022]
Abstract
Neonatal stroke is a leading cause of long-term disability and currently available rehabilitation treatments are insufficient to promote recovery. Activating neural precursor cells (NPCs) in adult rodents, in combination with rehabilitation, can accelerate functional recovery following stroke. Here, we describe a novel method of constraint-induced movement therapy (CIMT) in a rodent model of neonatal stroke that leads to improved functional outcomes, and we asked whether the recovery was correlated with expansion of NPCs. A hypoxia/ischemia (H/I) injury was induced on postnatal day 8 (PND8) via unilateral carotid artery ligation followed by systemic hypoxia. One week and two weeks post-H/I, CIMT was administered in the form of 3 botulinum toxin (Botox) injections, which induced temporary paralysis in the unaffected limb. Functional recovery was assessed using the foot fault task. NPC proliferation was assessed using the neurosphere assay and EdU immunohistochemistry. We found that neonatal H/I injury alone expands the NPC pool by >2.5-fold relative to controls. We determined that using Botox injections as a method to provide CIMT results in significant functional motor recovery after H/I. However, CIMT does not lead to enhanced NPC activation or migration into the injured parenchyma in vivo. At the time of functional recovery, increased numbers of proliferating inflammatory cells were found within the injured motor cortex. Together, these findings suggest that NPC activation following CIMT does not account for the observed functional improvement and suggests that CIMT-mediated modification of the CNS inflammatory response may play a role in the motor recovery.
Collapse
Affiliation(s)
- Kelsey V Adams
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Neemat Mahmud
- Department of Surgery, Division of Anatomy, University of Toronto, Toronto, ON, Canada
| | | | - Ilan Vonderwalde
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Daisuke Umebayashi
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Nadia Sachewsky
- Department of Surgery, Division of Anatomy, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Brenda L Coles
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Derek van der Kooy
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Cindi M Morshead
- Department of Surgery, Division of Anatomy, University of Toronto, Toronto, ON, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada.,KITE, Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
15
|
A novel compound DBZ ameliorates neuroinflammation in LPS-stimulated microglia and ischemic stroke rats: Role of Akt(Ser473)/GSK3β(Ser9)-mediated Nrf2 activation. Redox Biol 2020; 36:101644. [PMID: 32863210 PMCID: PMC7371982 DOI: 10.1016/j.redox.2020.101644] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/30/2020] [Accepted: 07/11/2020] [Indexed: 01/07/2023] Open
Abstract
Microglia-mediated neuroinflammation plays a crucial role in the pathophysiological process of multiple neurological disorders such as ischemic stroke, yet lacks effective therapeutic agents. Previously, we discovered one novel synthetic compound, tanshinol borneol ester (DBZ), possesses anti-inflammatory and anti-atherosclerotic activities, whereas little is known about its effects in CNS. Therefore, the present study aims to explore the effects and potential mechanism of DBZ on neuroinflammation and microglial function. Our studies revealed that DBZ significantly inhibited NF-κB activity, suppressed the production of pro-inflammatory mediators meanwhile promoted M2 mediators expression in LPS-stimulated BV2 cells and mouse primary microglia cells. DBZ also exhibited antioxidant activity by enhancing Nrf2 nuclear accumulation and transcriptional activity, increasing HO-1 and NQO1 expression, and inhibiting LPS-induced ROS generation in BV2 cells. Importantly, the anti-neuroinflammatory and antioxidant effects of DBZ above were reversed by Nrf2 knockdown. Additionally, DBZ ameliorated sickness behaviors of neuroinflammatory mice induced by systemic LPS administration, and significantly reduced infract volume, improved sensorimotor and cognitive function in rats subjected to transient middle cerebral artery occlusion (tMCAO); besides, DBZ restored microglia morphological alterations and shifted the M1/M2 polarization in both murine models. Mechanistically, DBZ-induced Nrf2 nuclear accumulation and antioxidant enzymes expression were accompanied by increased level of p-Akt(Ser473) (activation) and p-GSK3β(Ser9) (inactivation), and decreased nuclear level of Fyn both in vitro and in vivo. Pharmacologically inhibiting PI3K or activating GSK3β markedly increased nuclear density of Fyn in microglia cells, which blocked the promoting effect of DBZ on Nrf2 nuclear accumulation and its antioxidant and anti-neuroinflammatory activities. Collectively, these results indicated the effects of DBZ on microglia-mediated neuroinflammation were strongly associated with the nuclear accumulation and stabilization of Nrf2 via the Akt(Ser473)/GSK3β(Ser9)/Fyn pathway. With anti-neuroinflammatory and antioxidant properties, DBZ could be a promising new drug candidate for prevention and/or treatment of cerebral ischemia and other neuroinflammatory disorders.
Collapse
|
16
|
Kumar AJ, Martins DO, Arruda BP, Lee VY, Chacur M, Nogueira MI. Impairment of nociceptive responses after neonatal anoxia correlates with somatosensory thalamic damage: A study in rats. Behav Brain Res 2020; 390:112690. [DOI: 10.1016/j.bbr.2020.112690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 10/24/2022]
|
17
|
Liu D, Bai X, Ma W, Xin D, Chu X, Yuan H, Qiu J, Ke H, Yin S, Chen W, Wang Z. Purmorphamine Attenuates Neuro-Inflammation and Synaptic Impairments After Hypoxic-Ischemic Injury in Neonatal Mice via Shh Signaling. Front Pharmacol 2020; 11:204. [PMID: 32194421 PMCID: PMC7064623 DOI: 10.3389/fphar.2020.00204] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 02/14/2020] [Indexed: 01/05/2023] Open
Abstract
Purmorphamine (PUR), an agonist of the Smoothened (Smo) receptor, has been shown to function as a neuroprotectant in acute experimental ischemic stroke. Its role in hypoxic-ischemic (HI) brain injury in neonatal mice remains unknown. Here we show that PUR attenuated acute brain injury, with a decrease in Bax/Bcl-2 ratio as well as inhibition of caspase-3 activation. These beneficial effects of PUR were associated with suppressing neuro-inflammation and oxidative stress. PUR exerted long-term protective effects upon tissue loss and improved neurobehavioral outcomes as determined at 14 and 28 days post-HI insult. Moreover, PUR increased synaptophysin (Syn) and postsynaptic density (PSD) protein 95 expression in HI-treated mice and attenuated synaptic loss. PUR upregulated the expression of Shh pathway mediators, while suppression of the Shh signaling pathway with cyclopamine (Cyc) reversed these beneficial effects of PUR on HI insult. Our study suggests a therapeutic potential for short-term PUR administration in HI-induced injury as a result of its capacity to exert multiple protective actions upon acute brain injury, long-term memory deficits, and impaired synapses. Moreover, we provide evidence indicating that one of the mechanisms underlying these beneficial effects of PUR involves activation of the Shh signaling pathway.
Collapse
Affiliation(s)
- Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Shandong University, Jinan, China
| | - Xuemei Bai
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Weiwei Ma
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Shandong University, Jinan, China.,Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Danqing Xin
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xili Chu
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Hongtao Yuan
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jie Qiu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Shandong University, Jinan, China.,Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - HongFei Ke
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Sen Yin
- Qilu Hospital, Shandong University, Jinan, China
| | | | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| |
Collapse
|
18
|
Kaur H, Xu N, Doycheva DM, Malaguit J, Tang J, Zhang JH. Recombinant Slit2 attenuates neuronal apoptosis via the Robo1-srGAP1 pathway in a rat model of neonatal HIE. Neuropharmacology 2019; 158:107727. [PMID: 31356825 PMCID: PMC6745244 DOI: 10.1016/j.neuropharm.2019.107727] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/02/2019] [Accepted: 07/25/2019] [Indexed: 12/21/2022]
Abstract
Apoptosis following hypoxic-ischemic injury to the brain plays a major role in neuronal cell death. The neonatal brain is more susceptible to injury as the cortical neurons are immature and there are lower levels of antioxidants. Slit2, an extracellular matrix protein, has been shown to be neuroprotective in various models of neurological diseases. However, there is no information about the role of Slit2 in neonatal hypoxia-ischemia. In this study, we evaluated the effect of Slit2 and its receptor Robo1 in a rat model with neonatal HIE. 10-day old rat pups were used to create the neonatal HIE model. The right common carotid artery was ligated followed by 2.5 h of hypoxia. Recombinant Slit2 was administered intranasally 1 h post HI, recombinant Robo1 was used as a decoy receptor and administered intranasally 1h before HI and srGAP1-siRNA was administered intracerebroventricularly 24 h before HI. Brain infarct area measurement, short-term and long-term neurological function tests, Western blot, immunofluorescence staining, Fluoro-Jade C staining, Nissl staining and TUNEL staining were the assessments done following drug administration. Recombinant Slit2 administration reduced neuronal apoptosis and neurological deficits after neonatal HIE which were reversed by co-administration of recombinant Robo1 and srGAP1-siRNA administration. Recombinant Slit2 showed improved outcomes possibly via the robo1-srGAP1 pathway which mediated the inhibition of RhoA. In this study, the results suggest that Slit2 may help in attenuation of apoptosis and could be a therapeutic agent for treatment of neonatal hypoxic ischemic encephalopathy.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Ningbo Xu
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Desislava Met Doycheva
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Jay Malaguit
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA; Department of Anesthesiology, Neurosurgery and Neurology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
19
|
Halis H, Bitiktaş S, Baştuğ O, Tan B, Kavraal Ş, Güneş T, Süer C. Differential Effects of Pentoxifylline on Learning and Memory Impairment Induced by Hypoxic-ischemic Brain Injury in Rats. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2019; 17:388-399. [PMID: 31352705 PMCID: PMC6705102 DOI: 10.9758/cpn.2019.17.3.388] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 01/10/2023]
Abstract
Objective Hypoxic-ischemic (HI) brain injury in the human perinatal period often leads to significant long-term neurobehavioral dysfunction in the cognitive and sensory-motor domains. Using a neonatal HI injury model (unilateral carotid ligation followed by hypoxia) in postnatal day seven rats, the present study investigated the long-term effects of HI and potential behavioral protective effect of pentoxifylline. Methods Seven-day-old rats underwent right carotid ligation, followed by hypoxia (FiO2 = 0.08). Rats received pentoxifylline immediately after and again 2 hours after hypoxia (two doses, 60‒100 mg/kg/dose), or serum physiologic. Another set of seven-day-old rats was included to sham group exposed to surgical stress but not ligated. These rats were tested for spatial learning and memory on the simple place task in the Morris water maze from postnatal days 77 to 85. Results HI rats displayed significant tissue loss in the right hippocampus, as well as severe spatial memory deficits. Low-dose treatment with pentoxifylline resulted in significant protection against both HI-induced hippocampus tissue losses and spatial memory impairments. Beneficial effects are, however, negated if pentoxifylline is administered at high dose. Conclusion These findings indicate that unilateral HI brain injury in a neonatal rodent model is associated with cognitive deficits, and that low dose pentoxifylline treatment is protective against spatial memory impairment.
Collapse
Affiliation(s)
- Hülya Halis
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Soner Bitiktaş
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Osman Baştuğ
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Burak Tan
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Şehrazat Kavraal
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Tamer Güneş
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Cem Süer
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
20
|
Knox-Concepcion KR, Figueroa JD, Hartman RE, Li Y, Zhang L. Repression of the Glucocorticoid Receptor Increases Hypoxic-Ischemic Brain Injury in the Male Neonatal Rat. Int J Mol Sci 2019; 20:ijms20143493. [PMID: 31315247 PMCID: PMC6678481 DOI: 10.3390/ijms20143493] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) resulting from asphyxia is the most common cause of neonatal brain damage and results in significant neurological sequelae, including cerebral palsy. The current therapeutic interventions are extremely limited in improving neonatal outcomes. The present study tests the hypothesis that the suppression of endogenous glucocorticoid receptors (GRs) in the brain increases hypoxic-ischemic (HI) induced neonatal brain injury and worsens neurobehavioral outcomes through the promotion of increased inflammation. A mild HI treatment of P9 rat pups with ligation of the right common carotid artery followed by the treatment of 8% O2 for 60 min produced more significant brain injury with larger infarct size in female than male pups. Intracerebroventricular injection of GR siRNAs significantly reduced GR protein and mRNA abundance in the neonatal brain. Knockdown of endogenous brain GRs significantly increased brain infarct size after HI injury in male, but not female, rat pups. Moreover, GR repression resulted in a significant increase in inflammatory cytokines TNF-α and IL-10 at 6 h after HI injury in male pups. Male pups treated with GR siRNAs showed a significantly worsened reflex response and exhibited significant gait disturbances. The present study demonstrates that endogenous brain GRs play an important role in protecting the neonatal brain from HI induced injury in male pups, and suggests a potential role of glucocorticoids in sex differential treatment of HIE in the neonate.
Collapse
Affiliation(s)
- Katherine R Knox-Concepcion
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Johnny D Figueroa
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Richard E Hartman
- Department of Psychology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Yong Li
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
21
|
Aghoghovwia BE, Goddard L, Oorschot DE. Long-Term Fine Motor Capability on the Staircase Test Correlates with the Absolute Number, but Not the Density, of DARPP-Positive Neurons in the Caudate-Putamen. Anat Rec (Hoboken) 2019; 302:2040-2048. [PMID: 31177619 DOI: 10.1002/ar.24196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 12/03/2018] [Accepted: 01/07/2019] [Indexed: 11/08/2022]
Abstract
Measurement of long-term functional and anatomical outcomes in the same animal is considered a powerful strategy for correlating structure with function. In a neonatal animal model of hypoxic-ischemic brain injury that is relevant to cerebral palsy, long-term functional deficits on the staircase test and long-term anatomical deficits in the absolute number of medium-spiny projection neurons in the caudate-putamen were reported in different animals due to logistical constraints. Here, we investigated if these functional and anatomical measures were correlated when measured in the same animals. The medium-spiny projection neurons were investigated because (1) they comprise the vast majority (>97%) of all neurons in the caudate-putamen and (2) motor deficits observed during staircase testing are likely to involve these striatal medium-spiny projection neurons through their connections. We found that long-term skilled forepaw capability on the staircase test was correlated with the absolute number of DARPP-32-positive medium-spiny projection neurons in the caudate-putamen. Specifically, deficits in skilled forepaw ability for the number of sugar pellets eaten and retrieved, and for the maximum staircase level reached, were significantly correlated with a lower absolute neuronal number. We also found that skilled forepaw ability on the staircase test was not correlated with the neuronal density (i.e., number per unit volume) of DARPP-32-positive medium-spiny projection neurons. Since neuronal density is an indirect measure of neuronal survival that is used in the literature, and absolute neuronal number is a direct measure, the results also highlight the scientific value of measuring absolute neuronal number. Anat Rec, 302:2040-2048, 2019. © 2019 American Association for Anatomy.
Collapse
Affiliation(s)
- Benjamin E Aghoghovwia
- Department of Anatomy, School of Biomedical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Liping Goddard
- Department of Anatomy, School of Biomedical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Dorothy E Oorschot
- Department of Anatomy, School of Biomedical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| |
Collapse
|
22
|
Narang R, Carter K, Muncie C, Pang Y, Fan LWW, Feng Y, Ojeda NB, Bhatt AJ. Intrauterine growth restriction and neonatal hypoxic ischemic brain injury causes sex-specific long-term neurobehavioral abnormalities in rats. J Neurosci Res 2019; 97:661-672. [PMID: 30843634 DOI: 10.1002/jnr.24389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 12/28/2018] [Accepted: 01/02/2019] [Indexed: 12/22/2022]
Abstract
There is a lack of knowledge of factors preventing an adequate response to moderate hypothermia after hypoxic ischemic (HI) brain injury. We hypothesized that growth restriction from reduced intrauterine perfusion would predispose neonatal rats to have a worse outcome with HI brain injury. IUGR was induced by placental insufficiency in dams at 14 days of gestation. HI was induced at postnatal day (P) 10 by permanent right carotid artery ligation followed by 90 min of hypoxia (8% oxygen). Tests for early brain injury and neurobehavioral outcomes were subsequently done. All statistical analysis was done using Two-way ANOVA; post hoc Holm-Sidak test. HI in control and IUGR groups decreased the success rate of the contralateral vibrissa-elicited forelimb test, increased response latency in movement initiation test and increased the time to finish elevated beam walk test at P40 and P60. IUGR augmented HI-induced abnormality in vibrissa-elicited forelimb test at P40 but showed higher success rate when compared to HI only group at P60. IUGR's negative effect on HI-induced changes on the elevated beam walk test was sex-specific and exaggerated in P60 males. Increased TUNEL positive cells in the cortex were noted at 72 h after in HI in control but not in IUGR groups. In conclusion, the consequences of IUGR on subsequent neonatal HI varied based on age, sex and outcomes examined, and overall, male sex and IUGR had worse effects on the long-term neurobehavioral outcomes following HI.
Collapse
Affiliation(s)
- Radhika Narang
- Division of Newborn Medicine, Valley Children's Healthcare, Madera, California
| | - Kathleen Carter
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Colin Muncie
- Department of Surgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yi Pang
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lir-Wan W Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yangzheng Feng
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Norma B Ojeda
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Abhay J Bhatt
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
23
|
Muntsant A, Shrivastava K, Recasens M, Giménez-Llort L. Severe Perinatal Hypoxic-Ischemic Brain Injury Induces Long-Term Sensorimotor Deficits, Anxiety-Like Behaviors and Cognitive Impairment in a Sex-, Age- and Task-Selective Manner in C57BL/6 Mice but Can Be Modulated by Neonatal Handling. Front Behav Neurosci 2019; 13:7. [PMID: 30814939 PMCID: PMC6381068 DOI: 10.3389/fnbeh.2019.00007] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/09/2019] [Indexed: 12/13/2022] Open
Abstract
Perinatal brain injury (PBI) leads to neurological disabilities throughout life, from motor deficits, cognitive limitations to severe cerebral palsy. Yet, perinatal brain damage has limited therapeutic outcomes. Besides, the immature brain of premature children is at increased risk of hypoxic/ischemic (HI) injury, with males being more susceptible to it and less responsive to protective/therapeutical interventions. Here, we model in male and female C57BL/6 mice, the impact of neonatal HI and the protective effects of neonatal handling (NH), an early life tactile and proprioceptive sensory stimulation. From postnatal day 1 (PND1, modeling pre-term) to PND21 randomized litters received either NH or left undisturbed. HI brain damage occurred by permanent left carotid occlusion followed by hypoxia at PND7 (modeling full-term) in half of the animals. The behavioral and functional screening of the pups at weaning (PND23) and their long-term outcomes (adulthood, PND70) were evaluated in a longitudinal study, as follows: somatic development (weight), sensorimotor functions (reflexes, rods and hanger tests), exploration [activity (ACT) and open-field (OF) test], emotional and anxiety-like behaviors [corner, open-field and dark-light box (DLB) tests], learning and memory [T-maze (TM) and Morris Water-Maze (MWM)]. HI induced similar brain damage in both sexes but affected motor development, sensorimotor functions, induced hyperactivity at weaning, and anxiety-like behaviors and cognitive deficits at adulthood, in a sex- and age-dependent manner. Thus, during ontogeny, HI affected equilibrium especially in females and prehensility in males, but only reflexes at adulthood. Hyperactivity of HI males was normalized at adulthood. HI increased neophobia and other anxiety-like behaviors in males but emotionality in females. Both sexes showed worse short/long-term learning, but memory was more affected in males. Striking neuroprotective effects of NH were found, with significantly lower injury scores, mostly in HI males. At the functional level, NH reversed the impaired reflex responses and improved memory performances in hippocampal-dependent spatial-learning tasks, especially in males. Finally, neuropathological correlates referred to atrophy, neuronal densities and cellularity in the affected areas [hippocampal-CA, caudate/putamen, thalamus, neocortex and corpus callosum (CC)] point out distinct neuronal substrates underlying the sex- and age- functional impacts of these risk/protection interventions on sensorimotor, behavioral and cognitive outcomes from ontogeny to adulthood.
Collapse
Affiliation(s)
- Aida Muntsant
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Kalpana Shrivastava
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology & Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mireia Recasens
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology & Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lydia Giménez-Llort
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
24
|
Wright JL, Chu HX, Kagan BJ, Ermine CM, Kauhausen JA, Parish CL, Sobey CG, Thompson LH. Local Injection of Endothelin-1 in the Early Neonatal Rat Brain Models Ischemic Damage Associated with Motor Impairment and Diffuse Loss in Brain Volume. Neuroscience 2018; 393:110-122. [PMID: 30300704 DOI: 10.1016/j.neuroscience.2018.09.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/23/2018] [Accepted: 09/28/2018] [Indexed: 11/16/2022]
Abstract
Cerebral palsy is an irreversible movement disorder resulting from cerebral damage sustained during prenatal or neonatal brain development. As survival outcomes for preterm injury improve, there is increasing need to model ischemic injury at earlier neonatal time-points to better understand the subsequent pathological consequences. Here we demonstrate a novel neonatal ischemic model using focal administration of the potent vasoconstrictor peptide, endothelin-1 (ET-1), in newborn rats. The functional and histopathological outcomes compare favourably to those reported following the widely used hypoxic ischemia (HI) model. These include a robust motor deficit sustained into adulthood and recapitulation of hallmark features of preterm human brain injury, including atrophy of subcortical white matter and periventricular fiber bundles. Compared to procedures involving carotid artery manipulation and periods of hypoxia, the ET-1 ischemia model represents a rapid and technically simplified model more amenable to larger cohorts and with the potential to direct the locus of ischemic damage to specific brain areas.
Collapse
Affiliation(s)
- Jordan L Wright
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
| | - Hannah X Chu
- Biomedicine Discovery Institute and Department of Pharmocology, Monash University, Melbourne, VIC, Australia
| | - Brett J Kagan
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Charlotte M Ermine
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Jessica A Kauhausen
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Christopher G Sobey
- Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC, Australia
| | - Lachlan H Thompson
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
| |
Collapse
|
25
|
Mason B, Rollins LG, Asumadu E, Cange C, Walton N, Donaldson ST. Nesting Environment Provides Sex-Specific Neuroprotection in a Rat Model of Neonatal Hypoxic-Ischemic Injury. Front Behav Neurosci 2018; 12:221. [PMID: 30356904 PMCID: PMC6190890 DOI: 10.3389/fnbeh.2018.00221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 09/03/2018] [Indexed: 11/17/2022] Open
Abstract
Hypoxic-ischemic (HI) encephalopathy is a devastating injury that occurs when the fetal brain is deprived of oxygen and blood to a degree that may lead to neurological damage, seizing and cerebral palsy. In rodents, early environmental enrichment that promotes maternal care-taking behavior (mCTB) can improve neurobehavioral outcomes and protect against neurological decline. We hypothesized that an enhanced nesting environment would improve mCTB as measured by pup weight gain, and support greater HI recovery in developing rats. Pregnant dams (E15-16) were introduced to either control Standard Facility (SF) housing or closed nestbox (CN) conditions and maintained in larger cages through pup weaning. On postnatal day (PND) 7, male and female Long-Evans rat pups (N = 73) were randomly sorted into one of two surgical conditions: control and HI. HI pups received isoflurane anesthesia and right carotid artery ligation, a 2-h rest followed by 90 min exposure to a moist hypoxic (92% N, 8% O2) chamber. Pups (PND 8) were weighed daily, and tested on the Morris Water Maze (MWM) task (PND 35-50). Results demonstrate significant differences afforded to male and female pups based on weight measure, where CN-rearing modifies pre-weaning adolescent weights in females and increases post-weaning weights in males and females by an average of 10 g. Following successful MWM training and acquisition (PND 35-37), both male and female CN-raised animals demonstrated faster latency to find the hidden platform (HP) during HP trials (PND 38-42) and appeared to freely explore the MWM pool during an additional probe trial (PND 43). Moreover, after sacrifice (PND 60), CN rearing created sex-specific alterations in brain-derived neurotrophic factor (BDNF), glial-derived neurotrophic factor (GDNF) immunopositive cell staining of the dorsomedial striatum and CA1 of the hippocampus. CN-rearing afforded HI males higher BDNF levels in the striatum and produced greater GDNF levels in the hippocampus of HI-injured females. These results suggest that early life environmental enrichment positively modifies nesting environment, increases weight gain, as well as spatial learning and memory in a sex-specific directionality. Our findings also implicate correlative changes in corticolimbic neurotrophin protein levels in the CN-reared animals that may contribute to these benefits.
Collapse
Affiliation(s)
- Briana Mason
- Developmental and Brain Sciences, Department of Psychology, University of Massachusetts Boston, Boston, MA, United States
| | - L. G. Rollins
- Clinical Psychology Program, Department of Psychology, University of Massachusetts Boston, Boston, MA, United States
- Warren Alpert Medical School, Department of Psychiatry, Brown University, Providence, RI, United States
| | - Evans Asumadu
- Developmental and Brain Sciences, Department of Psychology, University of Massachusetts Boston, Boston, MA, United States
| | - Christina Cange
- Developmental and Brain Sciences, Department of Psychology, University of Massachusetts Boston, Boston, MA, United States
| | - Najah Walton
- Developmental and Brain Sciences, Department of Psychology, University of Massachusetts Boston, Boston, MA, United States
| | - S. Tiffany Donaldson
- Developmental and Brain Sciences, Department of Psychology, University of Massachusetts Boston, Boston, MA, United States
| |
Collapse
|
26
|
Xin D, Chu X, Bai X, Ma W, Yuan H, Qiu J, Liu C, Li T, Zhou X, Chen W, Liu D, Wang Z. l-Cysteine suppresses hypoxia-ischemia injury in neonatal mice by reducing glial activation, promoting autophagic flux and mediating synaptic modification via H 2S formation. Brain Behav Immun 2018; 73:222-234. [PMID: 29751053 DOI: 10.1016/j.bbi.2018.05.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/23/2018] [Accepted: 05/07/2018] [Indexed: 10/17/2022] Open
Abstract
We previously reported that l-Cysteine, an H2S donor, significantly alleviated brain injury after hypoxia-ischemic (HI) injury in neonatal mice. However, the mechanisms underlying this neuroprotective effect of l-Cysteine against HI insult remain unknown. In the present study, we tested the hypothesis that the protective effects of l-Cysteine are associated with glial responses and autophagy, and l-Cysteine attenuates synaptic injury as well as behavioral deficits resulting from HI. Consistent with our previous findings, we found that treatment with l-Cysteine after HI reduced early brain injury, improved behavioral deficits and synaptic damage, effects which were associated with an up-regulation of synaptophysin and postsynaptic density protein 95 expression in the lesioned cortex. l-Cysteine attenuated the accumulation of CD11b+/CD45high cells, activation of microglia and astrocytes and diminished HI-induced increases in reactive oxygen species and malondialdehyde within the lesioned cortex. In addition, l-Cysteine increased microtubule associated protein 1 light chain 3-II and Beclin1 expression, decreased p62 expression and phosphor-mammalian target of rapamycin and phosphor-signal transducer and activator of transcription 3. Further support for a critical role of l-Cysteine was revealed from results demonstrating that treatment with an inhibitor of the H2S-producing enzyme, amino-oxyacetic acid, reversed the beneficial effects of l-Cysteine described above. These results demonstrate that l-Cysteine effectively alleviates HI injury and improves behavioral outcomes by inhibiting reactive glial responses and synaptic damage and an accompanying triggering of autophagic flux. Accordingly, l-Cysteine may provide a new a therapeutic approach for the treatment of HI via the formation of H2S.
Collapse
Affiliation(s)
- Danqing Xin
- Department of Physiology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Xili Chu
- Department of Physiology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Xuemei Bai
- Department of Physiology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Weiwei Ma
- Department of Physiology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Hongtao Yuan
- Department of Physiology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China; Department of Medical Psychology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Jie Qiu
- Department of Physiology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China; Department of Medical Psychology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Changxing Liu
- Department of Physiology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Tong Li
- Department of Physiology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Xin Zhou
- Department of Physiology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Wenqiang Chen
- Shandong University Qilu Hospital, Jinan, Shandong, PR China
| | - Dexiang Liu
- Department of Medical Psychology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Zhen Wang
- Department of Physiology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China.
| |
Collapse
|
27
|
Long-term effects of enriched environment following neonatal hypoxia-ischemia on behavior, BDNF and synaptophysin levels in rat hippocampus: Effect of combined treatment with G-CSF. Brain Res 2017; 1667:55-67. [DOI: 10.1016/j.brainres.2017.05.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 04/13/2017] [Accepted: 05/01/2017] [Indexed: 12/12/2022]
|
28
|
Hypoxic postconditioning improves behavioural deficits at 6 weeks following hypoxic-ischemic brain injury in neonatal rats. Behav Brain Res 2017. [PMID: 28647597 DOI: 10.1016/j.bbr.2017.06.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxic-ischemic (HI) brain injury in newborns is associated with high morbidity and mortality, with many babies suffering neurological deficits. Recently, we showed that hypoxic postconditioning (PostC) immediately post injury can protect against HI up to one week in neonatal rats. Here, we aimed to examine whether long term functional deficits were also improved by PostC. Sprague-Dawley rats were assigned to control (C) or HI group on postnatal day 7 (P7). The HI group underwent unilateral carotid artery occlusion followed by hypoxia (7% oxygen, 3h). Half of each group were randomly assigned to the PostC group (8% oxygen, 1h/day for 5days post-injury), or normoxic group, where animals were kept under ambient conditions. Righting reflex and negative geotaxis tests were performed on P8 and P14. On P42, rats underwent further behavioural tests of motor function and memory (forelimb grip strength, grid walking and novel object recognition tasks). Brain injury was assessed using histological scoring of brain sections. At P14, PostC reduced the righting reflex deficit compared to HI alone. Long-term (6 weeks) behavioural deficits were observed in grid walking and novel object recognition tests after HI alone, with both functions improved following PostC. Following HI, there was an increase in brain injury assessed by histological scoring compared to control, and this damage was reduced by PostC. This novel finding of long-term histological neuroprotection accompanied by functional improvements by PostC further demonstrates the clinical potential of mild hypoxia for the treatment of HI brain injury.
Collapse
|
29
|
Cannabidiol reduces brain damage and improves functional recovery in a neonatal rat model of arterial ischemic stroke. Neuropharmacology 2017; 116:151-159. [DOI: 10.1016/j.neuropharm.2016.12.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/14/2016] [Accepted: 12/19/2016] [Indexed: 12/22/2022]
|
30
|
Netto CA, Sanches E, Odorcyk FK, Duran-Carabali LE, Weis SN. Sex-dependent consequences of neonatal brain hypoxia-ischemia in the rat. J Neurosci Res 2016; 95:409-421. [DOI: 10.1002/jnr.23828] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Carlos Alexandre Netto
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Eduardo Sanches
- Division of Child Development and Growth, Department of Pediatrics; University of Geneva; Geneva Switzerland
| | - Felipe Kawa Odorcyk
- Postgraduate Program of Neurosciences, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Luz Elena Duran-Carabali
- Postgraduate Program of Physiology, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Simone Nardin Weis
- Department of Cellular Biology; Universidade de Brasília; Brasilia Distrito Federal Brazil
| |
Collapse
|
31
|
Docosahexaenoic Acid Reduces Cerebral Damage and Ameliorates Long-Term Cognitive Impairments Caused by Neonatal Hypoxia-Ischemia in Rats. Mol Neurobiol 2016; 54:7137-7155. [PMID: 27796751 DOI: 10.1007/s12035-016-0221-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/12/2016] [Indexed: 10/20/2022]
Abstract
As the interest in the neuroprotective possibilities of docosahexaenoic acid (DHA) for brain injury has grown in the recent years, we aimed to investigate the long-term effects of this fatty acid in an experimental model of perinatal hypoxia-ischemia in rats. To this end, motor activity, aspects of learning, and memory function and anxiety, as well as corticofugal connections visualized by using tracer injections, were evaluated at adulthood. We found that in the hours immediately following the insult, DHA maintained mitochondrial inner membrane integrity and transmembrane potential, as well as the integrity of synaptic processes. Seven days later, morphological damage at the level of the middle hippocampus was reduced, since neurons and myelin were preserved and the astroglial reactive response and microglial activation were seen to be diminished. At adulthood, the behavioral tests revealed that treated animals presented better long-term working memory and less anxiety than non-treated hypoxic-ischemic animals, while no difference was found in the spontaneous locomotor activity. Interestingly, hypoxic-ischemic injury caused alterations in the anterograde corticofugal neuronal connections which were not so evident in rats treated with DHA. Thus, our results indicate that DHA treatment can lead to long-lasting neuroprotective effects in this experimental model of neonatal hypoxia-ischemic brain injury, not only by mitigating axonal changes but also by enhancing cognitive performance at adulthood.
Collapse
|
32
|
Studies on cerebral protection of digoxin against hypoxic–ischemic brain damage in neonatal rats. Neuroreport 2016; 27:906-15. [DOI: 10.1097/wnr.0000000000000630] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
33
|
Nafamostat mesilate improves function recovery after stroke by inhibiting neuroinflammation in rats. Brain Behav Immun 2016; 56:230-45. [PMID: 27033633 DOI: 10.1016/j.bbi.2016.03.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/10/2016] [Accepted: 03/23/2016] [Indexed: 01/20/2023] Open
Abstract
Inflammation plays an important role in stroke pathology, making it a promising target for stroke intervention. Nafamostat mesilate (NM), a wide-spectrum serine protease inhibitor, is commonly used for treating inflammatory diseases, such as pancreatitis. However, its effect on neuroinflammation after stroke was unknown. Hence, the effects of NM on the inflammatory response post stroke were characterized. After transient middle cerebral artery occlusion (tMCAO) in rats, NM reduced the infarct size, improved behavioral functions, decreased the expression of proinflammatory mediators (TNF-α, IL-1β, iNOS and COX-2) in a time-dependent manner and promoted the expression of different anti-inflammatory factors (CD206, TGF-β, IL-10 and IL-4) at different time points. Furthermore, NM could inhibit the expression of proinflammatory mediators and promote anti-inflammatory mediators expression in rat primary microglia following exposure to thrombin combined with oxygen-glucose deprivation (OGD). The immune-modulatory effect of NM might be partly due to its inhibition of the NF-κB signaling pathway and inflammasome activation after tMCAO. In addition, NM significantly inhibited the infiltration of macrophage, neutrophil and T lymphocytes, which was partly mediated by the inhibition of monocyte chemotactic protein-1 (MCP-1), intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). Taken together, our results indicated that NM can provide long-term protection of the brain against tMCAO by modulating a broad components of the inflammatory response.
Collapse
|
34
|
Liu Y, Li C, Wang J, Fang Y, Sun H, Tao X, Zhou XF, Liao H. Nafamostat Mesilate Improves Neurological Outcome and Axonal Regeneration after Stroke in Rats. Mol Neurobiol 2016; 54:4217-4231. [DOI: 10.1007/s12035-016-9999-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/14/2016] [Indexed: 08/24/2023]
|
35
|
Chen J, Chen YH, Lv HY, Chen LT. Effect of hyperbaric oxygen on lipid peroxidation and visual development in neonatal rats with hypoxia-ischemia brain damage. Biomed Rep 2016; 5:136-140. [PMID: 27347417 DOI: 10.3892/br.2016.673] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/19/2016] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the effect of hyperbaric oxygen (HBO) on lipid peroxidation and visual development in a neonatal rat model of hypoxic-ischemic brain damage (HIBD). The rat models of HIBD were established by delayed uterus dissection and were divided randomly into two groups (10 rats each): HIBD and HBO-treated HIBD (HIBD+HBO) group. Another 20 rats that underwent sham-surgery were also divided randomly into the HBO-treated and control groups. The rats that underwent HBO treatment received HBO (0.02 MPa, 1 h/day) 24 h after the surgery and this continued for 14 days. When rats were 4 weeks old, their flash visual evoked potentials (F-VEPs) were monitored and the ultrastructures of the hippocampus were observed under transmission electron microscope. The levels of superoxide dismutase (SOD) and malonyldialdehyde (MDA) in the brain tissue homogenate were detected by xanthine oxidase and the thiobarbituric acid colorimetric method. Compared with the control group, the ultrastructures of the pyramidal neurons in the hippocampal CA3 area were distorted, the latencies of F-VEPs were prolonged (P<0.01) and the SOD activities were lower while the MDA levels were higher (P<0.01) in the HIBD group. No significant differences in ultrastructure, the latency of F-VEPs or SOD/MDA levels were identified between the HBO-treated HIBD group and the normal control group (P>0.05). HBO enhances antioxidant capacity and reduces the ultrastructural damage induced by hypoxic-ischemia, which may improve synaptic reconstruction and alleviate immature brain damage to promote the habilitation of brain function.
Collapse
Affiliation(s)
- Jing Chen
- Department of Pediatrics, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China; Department of Child Health, Xiamen Maternal and Child Health Care Hospital, Xiamen, Fujian 361003, P.R. China
| | - Yan-Hui Chen
- Department of Pediatrics, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Hong-Yan Lv
- Department of Pediatrics, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Li-Ting Chen
- Department of Pediatrics, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
36
|
Rumajogee P, Bregman T, Miller SP, Yager JY, Fehlings MG. Rodent Hypoxia-Ischemia Models for Cerebral Palsy Research: A Systematic Review. Front Neurol 2016; 7:57. [PMID: 27199883 PMCID: PMC4843764 DOI: 10.3389/fneur.2016.00057] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 04/03/2016] [Indexed: 12/28/2022] Open
Abstract
Cerebral palsy (CP) is a complex multifactorial disorder, affecting approximately 2.5-3/1000 live term births, and up to 22/1000 prematurely born babies. CP results from injury to the developing brain incurred before, during, or after birth. The most common form of this condition, spastic CP, is primarily associated with injury to the cerebral cortex and subcortical white matter as well as the deep gray matter. The major etiological factors of spastic CP are hypoxia/ischemia (HI), occurring during the last third of pregnancy and around birth age. In addition, inflammation has been found to be an important factor contributing to brain injury, especially in term infants. Other factors, including genetics, are gaining importance. The classic Rice-Vannucci HI model (in which 7-day-old rat pups undergo unilateral ligation of the common carotid artery followed by exposure to 8% oxygen hypoxic air) is a model of neonatal stroke that has greatly contributed to CP research. In this model, brain damage resembles that observed in severe CP cases. This model, and its numerous adaptations, allows one to finely tune the injury parameters to mimic, and therefore study, many of the pathophysiological processes and conditions observed in human patients. Investigators can recreate the HI and inflammation, which cause brain damage and subsequent motor and cognitive deficits. This model further enables the examination of potential approaches to achieve neural repair and regeneration. In the present review, we compare and discuss the advantages, limitations, and the translational value for CP research of HI models of perinatal brain injury.
Collapse
Affiliation(s)
- Prakasham Rumajogee
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network , Toronto, ON , Canada
| | - Tatiana Bregman
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network , Toronto, ON , Canada
| | - Steven P Miller
- Department of Pediatrics, Hospital for Sick Children , Toronto, ON , Canada
| | - Jerome Y Yager
- Division of Pediatric Neurosciences, Stollery Children's Hospital, University of Alberta , Edmonton, AB , Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada; Division of Neurosurgery, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
37
|
Early environmental enrichment affects neurobehavioral development and prevents brain damage in rats submitted to neonatal hypoxia-ischemia. Neurosci Lett 2016; 617:101-7. [PMID: 26872850 DOI: 10.1016/j.neulet.2016.02.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 02/06/2016] [Accepted: 02/06/2016] [Indexed: 12/13/2022]
Abstract
Our previous results demonstrated improved cognition in adolescent rats housed in environmental enrichment (EE) that underwent neonatal hypoxia-ischemia (HI). The aim of this study was to investigate the effects of early EE on neurobehavioral development and brain damage in rats submitted to neonatal HI. Wistar rats were submitted to the HI procedure on the 7th postnatal day (PND) and housed in an enriched environment (8th-20th PND). The maturation of physical characteristics and the neurological reflexes were evaluated and the volume of striatum, corpus callosum and neocortex was measured. Data analysis demonstrated a clear effect of EE on neurobehavioral development; also, daily performance was improved in enriched rats on righting, negative geotaxis and cliff aversion reflex. HI caused a transient motor deficit on gait latency. Brain atrophy was found in HI animals and this damage was partially prevented by the EE. In conclusion, early EE stimulated neurobehavioral development in neonate rats and also protects the neocortex and the corpus callosum from atrophy following HI. These findings reinforce the potential of EE as a strategy for rehabilitation following neonatal HI and provide scientific support to the use of this therapeutic strategy in the treatment of neonatal brain injuries in humans.
Collapse
|
38
|
Yao Y, Zheng XR, Zhang SS, Wang X, Yu XH, Tan JL, Yang YJ. Transplantation of vascular endothelial growth factor-modified neural stem/progenitor cells promotes the recovery of neurological function following hypoxic-ischemic brain damage. Neural Regen Res 2016; 11:1456-1463. [PMID: 27857750 PMCID: PMC5090849 DOI: 10.4103/1673-5374.191220] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Neural stem/progenitor cell (NSC) transplantation has been shown to effectively improve neurological function in rats with hypoxic-ischemic brain damage. Vascular endothelial growth factor (VEGF) is a signaling protein that stimulates angiogenesis and improves neural regeneration. We hypothesized that transplantation of VEGF-transfected NSCs would alleviate hypoxic-ischemic brain damage in neonatal rats. We produced and transfected a recombinant lentiviral vector containing the VEGF165 gene into cultured NSCs. The transfected NSCs were transplanted into the left sensorimotor cortex of rats 3 days after hypoxic-ischemic brain damage. Compared with the NSCs group, VEGF mRNA and protein expression levels were increased in the transgene NSCs group, and learning and memory abilities were significantly improved at 30 days. Furthermore, histopathological changes were alleviated in these animals. Our findings indicate that transplantation of VEGF-transfected NSCs may facilitate the recovery of neurological function, and that its therapeutic effectiveness is better than that of unmodified NSCs.
Collapse
Affiliation(s)
- Yue Yao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiang-Rong Zheng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Shan-Shan Zhang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xia Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiao-He Yu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jie-Lu Tan
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yu-Jia Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
39
|
Zhang G, Chen L, Yang L, Hua X, Zhou B, Miao Z, Li J, Hu H, Namaka M, Kong J, Xu X. Combined use of spatial restraint stress and middle cerebral artery occlusion is a novel model of post-stroke depression in mice. Sci Rep 2015; 5:16751. [PMID: 26572587 PMCID: PMC4648085 DOI: 10.1038/srep16751] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 10/19/2015] [Indexed: 12/22/2022] Open
Abstract
Post stroke depression (PSD) is one of the most common complications of ischemic stroke. At present, the underlying mechanisms are unclear, largely because there are no reliable, valid and reproducible animal models of PSD. Here we report a novel animal model of PSD that displays consistent and reliable clinical features of hemiplegic stroke. The animal model encompasses a combination of the middle cerebral artery occlusion (MCAO) and spatial restraint stress. We found that a 60-minute MCAO followed by spatial restraint stress for 2 h daily for 2 to 4 weeks from the fourth day after MCAO induced PSD-like depressive phenotypes in mice. Importantly, the mice showed exacerbated deficits of neurological functions and decreased body weights, which were accompanied with reduced levels of brain derived neurotrophic factor and neurotransmitters including serotonin and dopamine. In addition, we identified increased levels of serum cortisol in our PSD mice. Finally, we found that mice with PSD were responsive to the tri-cyclic antidepressant imipramine as evidenced by their attenuated depressive behaviors, increased body weights, recovered brain serotonin levels, and decreased serum cortisol levels. This mouse model replicates multiple features of human post-stroke depression and thus provides a new model for the investigation of PSD.
Collapse
Affiliation(s)
- Gaocai Zhang
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China.,Institute of Neuroscience, Soochow University, Suzhou City, China
| | - Li Chen
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China
| | - Lingli Yang
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China
| | - Xiaodong Hua
- Department of Biochemistry, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Beiqun Zhou
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China.,Institute of Neuroscience, Soochow University, Suzhou City, China
| | - Zhigang Miao
- Institute of Neuroscience, Soochow University, Suzhou City, China
| | - Jizhen Li
- Department of Neurology, Suzhou Kowloon Hospital, 118 Wansheng Street, Suzhou City, China
| | - Hua Hu
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China
| | - Michael Namaka
- College of Pharmacy and Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xingshun Xu
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China.,Institute of Neuroscience, Soochow University, Suzhou City, China
| |
Collapse
|
40
|
Tan X, Chen Y, Li J, Li X, Miao Z, Xin N, Zhu J, Ge W, Feng Y, Xu X. The inhibition of Cdk5 activity after hypoxia/ischemia injury reduces infarct size and promotes functional recovery in neonatal rats. Neuroscience 2015; 290:552-60. [PMID: 25665755 DOI: 10.1016/j.neuroscience.2015.01.054] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 01/20/2015] [Accepted: 01/23/2015] [Indexed: 01/05/2023]
Abstract
Recent studies indicate that over-activation of Cdk5 is a crucial pro-death signal and Cdk5 activity inhibition provides neuroprotection in animal stroke models. However, Cdk5 inhibitors are reported to affect physiological functions of Cdk5 and lead to serious side effects. Therefore, targeting Cdk5 or its activators without affecting physiological functions of Cdk5 is a therapeutic strategy for ischemic brain injury. In this study, we examined Cdk5 activity in a rat hypoxia/ischemia (HI) injury model. Cdk5 expression was not changed after HI injury, but Cdk5 activity significantly increased, which was demonstrated by the increased phorsphorylation-phosphorylation of Tau and glucocorticoid receptor (GR), two downstream signals of Cdk5. We further showed that the levels of Cdk5 activators p35 and p39 decreased after HI injury, while p25, which is converted from p35 and has a higher activator activity on Cdk5, increased markedly after HI injury. P5, a 24-residue mimetic peptide of p35, was reported to specifically inhibit the p25/Cdk5 signal pathway in an Alzheimer's disease model. P5-TAT, which can cross the blood-brain barrier and cell membrane facilitated by TAT protein, was used in our study. We found that p5-TAT treatment did not change the levels of p35, p39, and p25, but reduced the phorsphorylation of Tau and GR, suggesting the inhibition of the p25/Cdk5 by the peptide p5-TAT. This was supported by the fact that p5 interacted with Cdk5, but not with Cdk5 activators. In addition, p5-TAT reduced cleaved caspase-3 level, a marker of neuronal apoptosis. We further demonstrated that p5-TAT pre-treatment reduced cerebral infarct volume; even when p5-TAT was delayed to be administered at 24h after HI injury, p5-TAT still promoted long-term functional recovery. Therefore, Cdk5 inhibition by the small peptide p5-TAT or its derivatives is a promising therapeutic strategy for the treatment of ischemic brain injury including hypoxic-ischemic encephalopathy and stroke.
Collapse
Affiliation(s)
- X Tan
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou City, Jiangsu Province, China; The Institute of Neuroscience, Soochow University, Suzhou City, Jiangsu Province, China
| | - Y Chen
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou City, Jiangsu Province, China; The Institute of Neuroscience, Soochow University, Suzhou City, Jiangsu Province, China
| | - J Li
- Department of Neurology, Suzhou Kowloon Hospital, 118 Wansheng Street, Suzhou City, China
| | - X Li
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou City, Jiangsu Province, China; The Institute of Neuroscience, Soochow University, Suzhou City, Jiangsu Province, China
| | - Z Miao
- The Institute of Neuroscience, Soochow University, Suzhou City, Jiangsu Province, China
| | - N Xin
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou City, Jiangsu Province, China; The Institute of Neuroscience, Soochow University, Suzhou City, Jiangsu Province, China
| | - J Zhu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou City, Jiangsu Province, China
| | - W Ge
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou City, Jiangsu Province, China
| | - Y Feng
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA.
| | - X Xu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou City, Jiangsu Province, China; The Institute of Neuroscience, Soochow University, Suzhou City, Jiangsu Province, China.
| |
Collapse
|
41
|
|
42
|
Zhao P, Ji G, Xue H, Yu W, Zhao X, Ding M, Yang Y, Zuo Z. Isoflurane postconditioning improved long-term neurological outcome possibly via inhibiting the mitochondrial permeability transition pore in neonatal rats after brain hypoxia–ischemia. Neuroscience 2014; 280:193-203. [DOI: 10.1016/j.neuroscience.2014.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 09/05/2014] [Accepted: 09/05/2014] [Indexed: 12/19/2022]
|
43
|
Chen J, Sun M, Zhang X, Miao Z, Chua BHL, Hamdy RC, Zhang QG, Liu CF, Xu X. Increased oligodendrogenesis by humanin promotes axonal remyelination and neurological recovery in hypoxic/ischemic brains. Hippocampus 2014; 25:62-71. [PMID: 25139533 DOI: 10.1002/hipo.22350] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2014] [Indexed: 12/12/2022]
Abstract
Oligodendrocytes are the predominant cell type in white matter and are highly vulnerable to ischemic injury. The role of oligodendrocyte dysfunction in ischemic brain injury is unknown. In this study, we used a 24-amino acid peptide S14G-Humanin (HNG) to examine oligodendrogenesis and neurological functional recovery in a hypoxic/ischemic (H/I) neonatal model. Intraperitoneal HNG pre-treatment decreased infarct volume following H/I injury. Delayed HNG treatment 24 h after H/I injury did not reduce infarct volume but did decrease neurological deficits and brain atrophy. Delayed HNG treatment did not attenuate axonal demyelination at 48 h after H/I injury. However, at 14 d after H/I injury, delayed HNG treatment increased axonal remyelination, the thickness of corpus callosum at the midline, the number of Olig2(+) /BrdU(+) cells, and levels of brain-derived neurotrophic factor (BDNF). Our results suggest that targeting oligodendrogenesis via delayed HNG treatment may represent a promising approach for the treatment of stroke.
Collapse
Affiliation(s)
- Jing Chen
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou City, China; The Institute of Neuroscience, Soochow University, Suzhou City, China
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Manaenko A, Lekic T, Barnhart M, Hartman R, Zhang JH. Inhibition of transforming growth factor-β attenuates brain injury and neurological deficits in a rat model of germinal matrix hemorrhage. Stroke 2014; 45:828-34. [PMID: 24425124 DOI: 10.1161/strokeaha.113.003754] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Transforming growth factor-β (TGF-β) overproduction and activation of the TGF-β pathway are associated with the development of brain injury following germinal matrix hemorrhage (GMH) in premature infants. We examined the effects of GMH on the level of TGF-β1 in a novel rat collagenase-induced GMH model and determined the effect of inhibition of the TGF receptor I. METHODS In total, 92 seven-day old (P7) rats were used. Time-dependent effects of GMH on the level of TGF-β1 and TGF receptor I were evaluated by Western blot. A TGF receptor I inhibitor (SD208) was administered daily for 3 days, starting either 1 hour or 3 days after GMH induction. The effects of GMH and SD208 on the TGF-β pathway were evaluated by Western blot at day 3. The effects of GMH and SD208 on cognitive and motor function were also assessed. The effects of TGF receptor I inhibition by SD208 on GMH-induced brain injury and underlying molecular pathways were investigated by Western blot, immunofluorescence, and morphology studies 24 days after GMH. RESULTS GMH induced significant delay in development, caused impairment in both cognitive and motor functions, and resulted in brain atrophy in rat subjects. GMH also caused deposition of both vitronectin (an extracellular matrix protein) and glial fibrillary acidic protein in perilesion areas, associated with development of hydrocephalus. SD208 ameliorated GMH-induced developmental delay, improved cognitive and motor functions, and attenuated body weight loss. SD208 also decreased vitronectin and glial fibrillary acidic protein deposition and decreased GMH-induced brain injury. CONCLUSIONS Increased level of TGF-β1 and activation of the TGF-β pathway associate with the development of brain injury after GMH. SD208 inhibits GMH-induced activation of the TGF-β pathway and leads to an improved developmental profile, partial recovery of cognitive and motor functions, and attenuation of GMH-induced brain atrophy and hydrocephalus.
Collapse
Affiliation(s)
- Anatol Manaenko
- From the Departments of Basic Science (A.M., T.L., J.H.Z.), Neurosurgery (J.H.Z.), Anesthesiology (M.B., J.H.Z.), and Psychology (R.H.), Loma Linda University, CA
| | | | | | | | | |
Collapse
|
45
|
Sullivan S, Friess SH, Ralston J, Smith C, Propert KJ, Rapp PE, Margulies SS. Improved behavior, motor, and cognition assessments in neonatal piglets. J Neurotrauma 2013; 30:1770-9. [PMID: 23758416 DOI: 10.1089/neu.2013.2913] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The alterations of animal behavior after traumatic brain injury (TBI) can be subtle, and their quantitative characterization can present significant methodological challenges. Meeting these challenges is a critical need, because quantitative measures are required in studies that compare the efficacy of different clinical interventions. We developed a battery of assessments to quantify behavioral, motor, and cognitive changes in neonatal piglets with good sensitivity and specificity to the detection of persistent deficits that correlate with axonal injury severity after a rapid non-impact head rotation with a diffuse pattern of axonal injury. The battery of measures developed included open field behaviors of sniffing and moving a toy, locomotion measures of Lempel-Ziv complexity and the probability of remaining in the current location, and a novel metric for evaluating motor performance. Our composite porcine disability score was able to detect brain injury with a sensitivity of 100% and specificity of 85.7% at day +4 post-injury for n=8 injured and n=7 sham piglets and significantly correlated with the percent axonal injury in these animals (day +4: ρ=0.76, p=0.0011). A significant improvement over our previous assessments, this new porcine disability score has potential use in a wide variety of porcine disease and injury models.
Collapse
Affiliation(s)
- Sarah Sullivan
- 1 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | | | | | | | | | | | | |
Collapse
|
46
|
Fang CZ, Yang YJ, Wang QH, Yao Y, Zhang XY, He XH. Intraventricular injection of human dental pulp stem cells improves hypoxic-ischemic brain damage in neonatal rats. PLoS One 2013; 8:e66748. [PMID: 23799131 PMCID: PMC3682969 DOI: 10.1371/journal.pone.0066748] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 05/10/2013] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To investigate the effect of intraventricular injection of human dental pulp stem cells (DPSCs) on hypoxic-ischemic brain damage (HIBD) in neonatal rats. METHODS Thirty-six neonatal rats (postnatal day 7) were assigned to control, HIBD, or HIBD+DPSC groups (n = 12 each group). For induction of HIBD, rats underwent left carotid artery ligation and were exposed to 8% to 10% oxygen for 2 h. Hoechst 33324-labeled human DPSCs were injected into the left lateral ventricle 3 days after HIBD. Behavioral assays were performed to assess hypoxic-ischemic encephalopathy (HIE), and on postnatal day 45, DPSC survival was assessed and expression of neural and glial markers was evaluated by immunohistochemistry and Western blot. RESULTS The HIBD group showed significant deficiencies compared to control on T-maze, radial water maze, and postural reflex tests, and the HIBD+DPSC group showed significant improvement on all behavioral tests. On postnatal day 45, Hoechst 33324-labeled DPSC nuclei were visible in the injected region and left cortex. Subsets of DPSCs showed immunostaining for neuronal (neuron-specific enolase [NSE], Nestin) and glial markers (glial fibrillary acidic protein [GFAP], O4). Significantly decreased staining/expression for NSE, GFAP, and O4 was found in the HBID group compared to control, and this was significantly increased in the HBID+DPSC group. CONCLUSION Intraventricular injection of human DPSCs improves HIBD in neonatal rats.
Collapse
Affiliation(s)
- Cheng-zhi Fang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Department of Neonatology, Wuhan Children's Hospital, Wuhan, China
| | - Yu-jia Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- * E-mail:
| | - Qin-hong Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yue Yao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-ying Zhang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Xue-hua He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
47
|
Rojas JJ, Deniz BF, Miguel PM, Diaz R, Hermel ÉDES, Achaval M, Netto CA, Pereira LO. Effects of daily environmental enrichment on behavior and dendritic spine density in hippocampus following neonatal hypoxia–ischemia in the rat. Exp Neurol 2013; 241:25-33. [DOI: 10.1016/j.expneurol.2012.11.026] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 11/26/2012] [Accepted: 11/29/2012] [Indexed: 11/24/2022]
|
48
|
Nayak PK, Kerr DS. Low-dose GYKI-52466: prophylactic preconditioning confers long-term neuroprotection and functional recovery following hypoxic-ischaemic brain injury. Neuroscience 2012; 232:128-38. [PMID: 23246617 DOI: 10.1016/j.neuroscience.2012.11.063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 11/28/2012] [Accepted: 11/29/2012] [Indexed: 10/27/2022]
Abstract
Experimental preconditioning provides beneficial outcomes in conditions such as cardiac surgery, brain surgery and stroke. Here we evaluated the protective effects of low-dose subcutaneous GYKI-52466 preconditioning in a rat model of hypoxic-ischaemic (HI) brain injury. Male Sprague-Dawley rats (postnatal day 26) were administered saline or GYKI-52466 (GYKI; 3-mg/kg, 90 min; 1-mg/kg, twice in 120 min; or 0.5-mg/kg, thrice in 180 min) prior to left common carotid artery occlusion. Animals were allowed to recover for 2h, and then placed in a hypoxia chamber (8% O₂/92% N₂; 33 ± 1°C) for 1h. A sham surgery group received saline without HI. Seizure activity was scored during hypoxia and sensorimotor tests performed before surgery and at 1, 7, 14 and 90 days post-HI. On days 14 and 90 brains were fixed and sectioned for the assessment of infarct size and ventricular enlargement. Low-dose GYKI-52466 preconditioning significantly reduced infarct volume and ventricular enlargement relative to saline-treated controls at day 14 after HI. On day 90, tissue loss was significantly reduced by GYKI 3-mg/kg compared to saline. Foot-faults, paw use asymmetry, and postural reflex scores were significantly improved in all GYKI treatment groups. Our results show that GYKI-52466 is effective at doses well-below, and at pre-administration intervals well-beyond previous studies, and suggest that a classical blockade of ionotropic AMPA receptors does not underlie its neuroprotective effects. Low-dose GYKI-52466 preconditioning represents a novel, prophylactic strategy for neuroprotection in a field almost devoid of effective pharmaceuticals.
Collapse
Affiliation(s)
- P K Nayak
- Department of Pharmacology and Toxicology, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | - D S Kerr
- Department of Pharmacology and Toxicology, University of Otago School of Medical Sciences, Dunedin, New Zealand.
| |
Collapse
|
49
|
Rosenkranz K, May C, Meier C, Marcus K. Proteomic analysis of alterations induced by perinatal hypoxic-ischemic brain injury. J Proteome Res 2012; 11:5794-803. [PMID: 23153068 DOI: 10.1021/pr3005869] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Perinatal hypoxic-ischemic brain injury is an important cause of neurological deficits still causing mortality and morbidity in the early period of life. As efficient clinical or pharmaceutical strategies to prevent or reduce the outcome of perinatal hypoxic-ischemic brain damage are limited, the development of new therapies is of utmost importance. To evolve innovative therapeutic concepts, elucidation of the mechanisms contributing to the neurological impairments upon hypoxic-ischemic brain injury is necessary. Therefore, we aimed for the identification of proteins that are affected by hypoxic-ischemic brain injury in neonatal rats. To assess changes in protein expression two days after induction of brain damage, a 2D-DIGE based proteome analysis was performed. Among the proteins altered after hypoxic-ischemic brain injury, Calcineurin A, Coronin-1A, as well as GFAP were identified, showing higher expression in lesioned hemispheres. Validation of the changes in Calcineurin A expression by Western Blot analysis demonstrated several truncated forms of this protein generated by limited proteolysis after hypoxia-ischemia. Further analysis revealed activation of calpain, which is involved in the limited proteolysis of Calcineurin. Active forms of Calcineurin are associated with the dephosphorylation of Darpp-32, an effect that was also demonstrated in lesioned hemispheres after perinatal brain injury.
Collapse
Affiliation(s)
- Katja Rosenkranz
- Department of Functional Proteomics, Ruhr-University Bochum, Germany.
| | | | | | | |
Collapse
|
50
|
Brima T, Mikulecká A, Otáhal J. Impacts of perinatal induced photothrombotic stroke on sensorimotor performance in adult rats. Physiol Res 2012; 62:85-94. [PMID: 23173682 DOI: 10.33549/physiolres.932447] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Perinatal ischemic stroke is a leading cerebrovascular disorder occurring in infants around the time of birth associated with long term comorbidities including motor, cognitive and behavioral deficits. We sought to determine the impact of perinatal induced stroke on locomotion, behavior and motor function in rats. A photothrombotic model of ischemic stroke was used in rat at postnatal day 7. Presently, we induced two lesions of different extents, to assess the consequences of stroke on motor function, locomotion and possible correlations to morphological changes. Behavioral tests sensitive to sensorimotor changes were used; locomotion expressed as distance moved in the open field was monitored and histological changes were also assessed. Outcomes depicted two kinds of lesions of different shapes and sizes, relative to laser illumination. Motor performance of rats submitted to stroke was poor when compared to controls; a difference in motor performance was also noted between rats with small and large lesions. Correlations were observed between: motor performance and exposition time; volume ratio and exposition time; and in the rotarod between motor performance and volume ratio. Outcomes demonstrate that photothrombotic cerebral ischemic stroke induced in early postnatal period and tested in adulthood, indeed influenced functional performance governed by the affected brain regions.
Collapse
Affiliation(s)
- T Brima
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | |
Collapse
|